EP2300003B1 - Compounds for use in treating injury associated with exposure to an alkylating species - Google Patents

Compounds for use in treating injury associated with exposure to an alkylating species Download PDF

Info

Publication number
EP2300003B1
EP2300003B1 EP09805313.5A EP09805313A EP2300003B1 EP 2300003 B1 EP2300003 B1 EP 2300003B1 EP 09805313 A EP09805313 A EP 09805313A EP 2300003 B1 EP2300003 B1 EP 2300003B1
Authority
EP
European Patent Office
Prior art keywords
cees
exposure
aeol
compound
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP09805313.5A
Other languages
German (de)
French (fr)
Other versions
EP2300003A2 (en
EP2300003A4 (en
Inventor
Brian J. Day
Carl W. White
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
National Jewish Health
Original Assignee
National Jewish Health
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by National Jewish Health filed Critical National Jewish Health
Priority to EP14154916.2A priority Critical patent/EP2732817B1/en
Publication of EP2300003A2 publication Critical patent/EP2300003A2/en
Publication of EP2300003A4 publication Critical patent/EP2300003A4/en
Application granted granted Critical
Publication of EP2300003B1 publication Critical patent/EP2300003B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41781,3-Diazoles not condensed 1,3-diazoles and containing further heterocyclic rings, e.g. pilocarpine, nitrofurantoin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/409Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil having four such rings, e.g. porphine derivatives, bilirubin, biliverdine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/06Free radical scavengers or antioxidants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • Sulfur mustards are classic vesicating agents that mainly affect the skin, eyes, and respiratory system. Medical surveillance of individuals exposed to mustard gas in the early 1980's has documented a number of respiratory conditions including bronchiolitis obliterans, asthma, and lung fibrosis that can persist throughout the victims' lifetime.
  • WO-A-02078670 discloses the treatment of patients who have suffered damage to the cerebrospinal tissue or who have risk of damage thereto comprising injecting a perfusion fluid in the cerebrospinal pathway containing a neuroprotectant, which may be a free radical inhibitor, including metalloporphyrins, including AEOL10150.
  • a neuroprotectant which may be a free radical inhibitor, including metalloporphyrins, including AEOL10150.
  • the damage can be ischemia, haemorrhage, trauma or exposure to toxic chemicals, such as chemical warfare agents, including the nerve agents VX and Sarin.
  • WO-A-02060383 describes that metalloporphyrins, including 10158, are useful for prevention of toxicity resulting from free radicals produced by chemotherapeutic agents, such as bleomycin, cisplatin, adriamycin (doxorubicin) and camptothicen.
  • chemotherapeutic agents such as bleomycin, cisplatin, adriamycin (doxorubicin) and camptothicen.
  • Disclosed herein are, inter alia, methods for rescuing or preventing organ injury following exposure to alkylating agents by using a substituted porphyrin as the active agent or alkylating agent protectant.
  • the methodology of the invention may implemented as follows.
  • a compound for use in treating an injury associated with exposure to an alkylating-agent, or for use in protecting a subject from the toxic effects associated with exposure to said alkylating agent the compound having the formula The above compound is also known as "AEOL 10150".
  • the injury may be associated with skin or lungs.
  • the alkylating agent may be a sulfur mustard or 2- chloroethyl ethyl sulfide.
  • the alkylating agent is a sulfur mustard.
  • Exposure to the alkylating agent may produce mitochondrial dysfunction, which in turn may result in an increase in reactive oxygen species production or oxidative stress.
  • exposure to the alkylating agent relative to non-exposure to the alkylating agent causes an increase in lactate dehydrogenase (LDH) levels, an increase in IgM levels, a decrease of glutathione levels, and an increase in myleperoxidase levels.
  • LDH lactate dehydrogenase
  • the compound may be administered by inhalation administration or topical administration.
  • the compound may be administered to the subject within about 0.5 hours to about 48 hours after exposure to the alkylating agent. More specifically, the compound may be administered to the subject within about 1 hour to about 10 hours after exposure to the alkylating agent.
  • Alkylating agent generally refers to compounds containing alkyl groups that readily attach to other molecules thereby forming a covalent bond. This process may also be referred to as alkylation. Generally, alkylating agents can disrupt DNA function through different mechanisms, such as: (i) by alkylating DNA bases, thereby preventing DNA synthesis and RNA transcription, (ii) by mediating the formation of cross-bridges, bonds between atoms in the DNA strand, or (iii) by facilitating the mispairing of the nucleotides in the DNA strand thereby leading to mutations.
  • alkylating agents may initiate oxidative stress within the cells of the exposed organ system causing an overall decrease in intracellular glutathione (GSH) and increased DNA oxidation. Exposure to alkylating agents may cause blistering of the skin, damage to the eyes, and damage to the respiratory tract. Exposure to alkylating agents may also cause systemic toxic effects, such as nausea and vomiting, reduction in both leukocytes and erythrocytes, hemorrhagic tendencies, edema, depletion of glutathione, increased myleperoxidase (MPO), increased lactate dehydrogenase (LDH), and increased IgM.
  • GSH intracellular glutathione
  • MPO myleperoxidase
  • LDH lactate dehydrogenase
  • Oxidation is a chemical reaction that transfers electrons from a substance to an oxidizing agent. Oxidation reactions may produce free radicals, which result in oxidative stress and may ultimately result in cell death.
  • Reactive oxygen species generally refers to free radicals, reactive anions containing oxygen atoms, or molecules containing oxygen atoms that can either produce free radicals or are chemically activated by them.
  • Reactive oxygen species may include, without limitation, superoxide radicals, hydrogen peroxide, peroxynitrite, lipid peroxides, hydroxyl radicals, thiyl radicals, superoxide anion, organic hydroperoxide, RO• alkoxy and ROO• peroxy radicals, and hypochlorous acid.
  • ROS reactive oxygen species
  • the main source of reactive oxygen species (ROS) in vivo is aerobic respiration, although reactive oxygen species are also produced by peroxisomal b-oxidation of fatty acids, microsomal cytochrome P450 metabolism of xenobiotic compounds, stimulation of phagocytosis by pathogens or lipopolysacchrides, arginine metabolism, tissue specific enzymes. Accumulating oxidative damage may also affect the efficiency of mitochondria and further increase the rate of ROS production.
  • Reactive nitrogen species generally refers to a family of biomolecules derived from nitric oxide (NO•) and may be produced in animals through the reaction of nitric oxide (NO•) with superoxide (O 2 - ) to form peroxynitrite (ONOO - ). In general, reactive nitrogen species act together with reactive oxygen species to damage cells, resulting in nitrosative stress.
  • Oxidative stress generally refers to cell damage caused by ROS.
  • the primary damage to cells results from the ROS-induced alteration of macromolecules such as polyunsaturated fatty acids in membrane lipids, essential proteins and DNA.
  • oxidative stress and ROS have been implicated in a number of disease states such as Alzheimer's disease, cancer, diabetes mellitus, and aging.
  • Antioxidant generally refers to molecules or compounds with the capability to attenuate or prevent the oxidation of other molecules. Antioxidants may remove free radicals generated from oxidation reaction and inhibit other oxidation reactions by becoming oxidized themselves. Antioxidants may include reducing agents such as thiols or polyphenols. Additionally, antioxidants may include, without limitation, glutathione, vitamin C, vitamin E, catalase, superoxide dismutase, glutathione peroxidase, various other peroxidases, the substituted porphyrin compounds of the invention and any other molecule or compound that is capable of scavenging reactive oxygen species known in the art.
  • Ratescue is generally defined as counteracting, recovering, or conferring protection from the deleterious effects of reactive oxygen species and other free radicals in a subject, organ, tissue, cell, or biomolecule.
  • Organ generally refers to a tissue that performs a specific function or group of functions within an organism.
  • An exemplary list of organs includes lungs, heart, blood vessels, blood, salivary glands, esophagus, stomach, liver, gallbladder, pancreas, intestines, rectum, anus, endocrine glands such as hypothalamus, pituitary or pituitary gland, pineal body or pineal gland, thyroid, parathyroids, adrenals, skin, hair, nails, lymph, lymph nodes, tonsils, adenoids, thymus, spleen, muscles, brain, spinal cord, peripheral nerves, nerves, sex organs such as ovaries, fallopian tubes, uterus, vagina, mammary glands, testes, vas deferens, seminal vesicles, prostate, and penis, pharynx, larynx, trachea, bronchi, diaphragm, bones
  • Organ system generally refers to a group of related organs. Organ systems include, without limitation, circulatory system, digestive system, endocrine system, integumentary system, lymphatic system, muscular system, nervous system, reproductive system, respiratory system, skeletal system, and urinary system.
  • Biomarker generally refers to an organic biomolecule which is differentially present in a sample taken from a subject of one phenotypic status (e.g., exposure to an alkylating agent) as compared with another phenotypic status (e.g., no exposure to an alkylating agent).
  • a biomarker is differentially present between different phenotypic statuses if the mean or median expression level of the biomarker in the different groups is calculated to be statistically significant. Common tests for statistical significance include, among others, t-test, ANOVA, Kruskal-Wallis, Wilcoxon, Mann-Whitney and odds ratio.
  • Biomarkers alone or in combination, provide measures of relative risk that a subject belongs to one phenotypic status or another. As such, they are useful as markers for disease (diagnostics), therapeutic effectiveness of a drug (theranostics), and for drug toxicity.
  • Subject includes individuals who require intervention or manipulation due to a exposure or potential exposure to an alkylating agent that can facilitate organ injury. Furthermore, the term “subject” includes non-human animals and humans.
  • Active agent generally refers to any compound capable of inducing a change in the phenotype or genotype of a cell, tissue, organ, or organism when contacted with the cell, tissue, organ, or organism.
  • the compound may have the ability to scavenge ROS, prevent or attenuate oxidative stress, and protect organs and organ systems from injury due to exposure to an alkylating agent.
  • the compound may include any substituted porphyrin compounds of the invention, such as a superoxide mimetic, a catalase mimetic or a mimetic having both features.
  • a “pharmaceutically acceptable carrier,” as used herein, generally refers to pharmaceutical excipients, for example, pharmaceutically, physiologically, acceptable organic or inorganic carrier substances suitable for enteral or parenteral application that do not deleteriously react with the active agent.
  • substituent groups are specified by their conventional chemical formulae, written from left to right, they equally encompass the chemically identical substituents that would result from writing the structure from right to left, e.g., -CH 2 O- is equivalent to -OCH 2 -.
  • Effective dose or “pharmaceutically effective dose,” as used herein, generally refers to the amount of the substituted porphyrin(s) described herein that produces a desired therapeutic effect, such as counteracting the deleterious effects of alkylating agent exposure.
  • the precise amount of the effective dose of a such a compound will yield the most effective results in terms of efficacy of treatment in a given subject will depend upon the activity, pharmacokinetics, pharmacodynamics, and bioavailability of a particular substituted porphyrin of the invention, physiological condition of the subject, the nature of the pharmaceutically acceptable carrier in a formulation, and a route of administration, among other potential factors.
  • methods are disclosed for treating, rescuing and/or protecting organ and organ systems in a subject from the deleterious effects resulting from exposure to alkylating agents using substituted porphyrins.
  • a method for treating an injury associated with exposure to an alkylating agent in a subject may include administering to a subject in need thereof an effective amount of AEOL10150.
  • a method for protecting a subject from the toxic effects associated with exposure to an alkylating agent may include administering prophylactically to a subject in need thereof an effective amount AEOL10150.
  • a method is disclosed for rescuing or protecting organ injury by administering AEOL10150 as the active agent of an alkylating agent protectant.
  • Compound AEOL 10150 is disclosed in U.S. Patent No. 7,189,707 and U.S. Patent Publication No. 2007/0149498 .
  • Alkylating agents contain alkyl groups that combine readily, typically through covalent bonding, with other molecules. Alkylating agents can disrupt DNA function by three mechanisms: (i) alkylating DNA bases, thereby preventing DNA synthesis and RNA transcription, (ii) mediating the formation of cross-bridges, bonds between atoms in the DNA strand, or (iii) facilitating the mis-pairing of the nucleotides in the DNA strand resulting in mutations in the DNA strand. Also, alkylating agents may initiate oxidative stress within the cells of the exposed organ system causing an overall decrease in intracellular glutathione (GSH) and increased DNA oxidation.
  • GSH glutathione
  • Sulfur mustard (2, 2'-dichloro diethyl sulfide) is a known potent vessicating agent and inhalation results in apoptosis and necrosis of the airway epithelium, inflammation, edema, and pseudomembrane formation.
  • 2-chloroethyl ethyl sulfide (CEES, half mustard) is a monofunctional analog of SM that can be utilized to elucidate the mechanisms of injury and as an initial screening of therapeutics.
  • Both SM and CEES ( Figure 1 ) are alkylating agents capable of binding macromolecules including proteins, DNA and lipids.
  • Oxidative stress plays a significant role in SM/CEES mediated damage.
  • exposure to CEES causes an imbalance in production of ROS/RNS and antioxidant defenses in favor of the former.
  • ROS/RNS ROS/RNS
  • antioxidant defenses There are many factors that contribute to the increase in ROS following SM/CEES exposure.
  • exposure to SM/CEES facilitates the proliferation of inflammatory cells such as polymorphonuclear leukocytes (PMN), which in turn produces oxidants, including superoxide and hypochlorous acid (HOCl).
  • PMN polymorphonuclear leukocytes
  • HOCl hypochlorous acid
  • exposure to CEES also results mitochrondrial dysfunction which further drives increased ROS production, and ultimately, oxidative stress.
  • AEOL 10150 has been shown to reduce alkylating agent-induced cytotoxicity, reduce alkylating agent-induced increases of protein and IgM in the lung, reduce levels of RBCs and inflammatory cells in the lung, decrease tissue accumulation of PMN, and prevent alkylating agent-induced oxidative stress.
  • the compound may be administered prophylactically to serve as a protectant against exposure to an alkylating agent.
  • the compound may be administered in the dosage amounts specified above about 1 hour to about 48 hours prior to alkylating agent exposure.
  • the compound of the invention may be administered about 1 to about 24 hours, more specifically, about 1 to about 12 hours, more specifically about 1 to about 6 hours, and even more specifically, about 1 to about 6 hours prior to alkylating agent exposure.
  • the therapeutically effective amount can be initially determined from cell culture assays.
  • Target concentrations will be those concentrations of active compound(s) that are capable of counteracting the effects of the alkylating agent, by monitoring the presence, absence, or alteration in levels of the biomarkers indicative of alkylating agent exposure, such as glutathione, LDH, IgM, and 80-HdG, for example. Methods for measuring the levels of such compounds is known by those of skill in the art and is matter of routine experimentation.
  • Therapeutically effective amounts for use in humans may be determined from animal models. For example, a dose for humans can be formulated to achieve a concentration that has been found to be effective in animals. The dosage in humans can be adjusted by monitoring the levels of the biomarkers indicative of exposure to an alkylating agent and adjusting the dosage upwards or downwards.
  • Table 1 Compound of Formula AEOL No. Designation IV AEOL 10153 V AEOL 10158 VI AEOL 10123 VII AEOL 10150 VIII AEOL 10151 IX AEOL 10303 X AEOL 10113 Compounds AEOL 10153, AEOL 10158, AEOL 10123, AEOL 10151, AEOL 10303 and AEOL 10113 are included in the examples for the purpose of reference and fall outside the scope of the claimed invention.
  • Human lung 16HBE cells were grown to approximately 90% confluence and treated with increasing concentrations of CEES, ranging from about 600 to about 000 ⁇ M. Cell viability was determined by measuring the fluorescence of calcein AM and was found to decrease in a dose-dependent manner from 80% with the 600 ⁇ M CEES to below 10% with 1000 ⁇ M CEES ( Figure 2 ). 900 ⁇ M CEES was used as the optimal dose to carry out the cytoprotection studies because it provided enough cell injury (about 50%) for potential therapeutics to demonstrate efficacy and the most consistent cell injury response in the two cell systems. Because of observed increased resistance of SAE cells to CEES toxicity as seen with 16HBE cells, these exposures were prolonged to 48 h in the SAE cells to provide similar injury responses for comparison of antioxidant protective effects between cell systems.
  • AEOL 10150 were assessed to determine whether its cytoprotective effects are associated with CEES-mediated changes in mitochondrial ROS and dysfunction.
  • Cells were grown to approximately 90% confluence and exposed to 900 ⁇ M CEES with and without AEOL 10150 (50 ⁇ M). Cells were incubated with MitoSOX 12 h after CEES exposure, and fluorescence was measured using flow cytometry.
  • AEOL 10150 added 1 h after CEES treatments significantly decreased mitochondrial ROS compared with CEES exposed cells in both SAE ( Figure 7A ) and 16HBE ( Figure 7B ) cells. AEOL 10150 alone did not cause a change in mitochondrial ROS.
  • AEOL 10150 was assessed to determine if it can protect the mitochondria from CEES-induced dysfunction.
  • Lung 16HBE cells were exposed to 900 ⁇ M CEES for 4 h with 50 ⁇ M AEOL 10150 added 1 h after the initial CEES exposure.
  • the CEES-only treated groups showed an increase in Rhodamine 123 fluorescence, indicating a significant loss of mitochondrial membrane potential that was attenuated in the AEOL 10150-treated cells ( Figure 7C ).
  • Oxidative stress can result from an imbalance between oxidant production and antioxidant defense.
  • GSH is a major cellular antioxidant. So, the effect of CEES on total cellular GSH levels was determined as well as whether AEOL 10150 altered CEES-mediated changes in GSH levels.
  • Human lung 16HBE cells were exposed for 12 h to CEES, and AEOL 10150 (50 ⁇ M) was added 1 h post-CEES treatment.
  • AEOL 10150 alone did not alter intracellular GSH levels, whereas CEES caused a significant decrease in intracellular GSH levels ( Figure 8A AEOL 10150 treatment prevented the CEES-induced decrease in GSH, further implicating an imbalance in redox status of the cells caused by CEES that was reversible by AEOL 10150.
  • oxidative stress is an increase in the oxidation of cellular macromolecules.
  • a classic marker for DNA oxidation is the formation of 8-hydroxydeoxyguanosine (8O-HdG), which was determined 12 h after CEES exposure.
  • CEES caused a significant increase in 80HdG levels in lung 16HBE cells as measured by high-performance liquid chromatography ( Figure 8B ).
  • AEOL 10150 added 1 h post-CEES exposure decreased CEES-mediated DNA oxidation.
  • Rats were exposed to 5% CEES for 15 minutes and killed 18 hours later. Groups of rats received AEOL 10150 (5 mg/kg sc, bid) 1 hour after CEES exposure. Rat lungs were lavaged and markers of cytotoxicity, inflammation and edema were measured in bronchoalveolar lavage fluid (BALF). As shown in Figure 9 , CEES caused a significant increase in the ROS. Moreover, AEOL 10150 added 1 h post-CEES exposure decreased CEES-mediated DNA oxidation. These data further support the role of oxidative stress in CEES-mediated injury that is ameliorated by the catalytic antioxidant metalloporphyrin, AEOL 10150.
  • CEES-induced cytotoxicity may be assessed by measuring LDH release in the lung.
  • LDH release in the bronchoalveolar lavage fluid (BAL) is a marker of cellular injury in the epithelium.
  • Figure 10 shows levels of LDH release were not different between EtOH + PBS and EtOH + AEOL 10150 treated animals. Following CEES exposure with PBS treatment, LDH release doubled as compared to the control groups (p ⁇ 0.01). When rats were administered AEOL 10150 following CEES-exposure, LDH levels were significantly attenuated as compared to the CEES + PBS group (p ⁇ 0.001).
  • Administering AEOL 10150 reduces alkylating agent-induced increases in protein and IgM in the lung.
  • BAL in normal rats consists of macrophages and low levels of large proteins such as albumin. Measuring protein levels in the BAL is one way to measure the accumulation of extravascular protein in the airways.
  • Figure 11A compared to EtOH + PBS or EtOH + AEOL 10150, protein levels in BAL were significantly increased as a result of 5% CEES + PBS ( p ⁇ 0.001). Protein levels in the BAL were significantly decreased from CEES + PBS when animals were administered AEOL 10150 (p ⁇ 0.001).
  • FIG. 11B demonstrates that IgM levels in the BAL were significantly increased a result in CEES + PBS rats as compared to EtOH + PBS or EtOH + AEOL 10150 (p ⁇ 0.001). IgM levels were significantly decreased with CEES + AEOL 10150 treatment as compared to CEES + PBS. Combined, these data demonstrate that administration of the AEOL 10150 following CEES exposure decreased protein levels in BAL as well as IgM levels.
  • Administering AEOL 10150 following alkylating agent exposure reduces levels of red blood cells (RBCs) and inflammatory cells in the lung. RBCs should not be present in the lung in any considerable levels unless there is hemorrhagic injury. Exposure to 5% CEES + PBS results in significantly increased hemorrhage as shown by increased RBC levels in the BAL ( p ⁇ 0.001). This CEES-induced damage is ameliorated with AEOL 10150 treatment 18 hours after CEES exposure (p ⁇ 0.05). Levels of PMN or neutrophils in the BAL were significantly increased in the CEES + PBS rats as compared to EtOH + PBS or EtOH+10150 (p ⁇ 0.001). CEES-induced neutrophil increases were significantly decreased with AEOL 10150 treatment (p ⁇ 0.05). While there was a decrease in macrophage levels with CEES exposure, this change did not reach significance as compared to the EtOH exposed animals.
  • MPO is a glycoprotein expressed in all cells of the myeloid lineage but is most abundant in the azurophilic granules ofPMNs. Released MPO by activated PMNs measured in whole lung homogenate demonstrates tissue accumulation and is a useful complement to measurement of PMN in the BAL. MPO levels were significantly increased as a result of CEES+PBS indicating an increase in PMN tissue accumulation (p ⁇ 0.01, Figure 12 ). AEOL 10150 treatment after CEES treatment significantly decreased tissue accumulation of PMN (p ⁇ 0.05).
  • Oxidative stress occurs when oxidant production exceeds antioxidant defense.
  • One marker of oxidative damage is DNA oxidation, which can be measured by the formation of 8-hydroxy-2-deoxyguanosine (8OHdG).
  • 8OHdG significantly increased in CEES+PBS rats as compared to levels in EtOH+PBS (p ⁇ 0.01) or EtOH+ 10150 ( p ⁇ 0.05) treatment 18 hours after exposure as measured by HPLC ( Figure 13 ).
  • 8O-HdG levels were significantly decreased as compared to CEES+PBS ( p ⁇ 0.05).
  • 4-HNE 4-hydroxynonenal
  • 4-HNE is a major product of total unsaturated aldehydes formed during lipid peroxidation. Measurement of 4-HNE levels in the lung 18 hours after CEES exposure resulted in a significant increase compared with EtOH+PBS treated rats ( Figure 14 ). AEOL 10150 significantly inhibited CEES- induced lipid peroxidation.

Description

    BACKGROUND OF THE INVENTION
  • Bis (2-chloroethyl sulfide) or sulfur mustard (SM) was first synthesized in the late 1880s and since has been used as a warfare agent on a number of occasions. SM was first used in World War I and has been used in warfare as recently as the Iran-Iraq conflict of the late 1980s. Although SM is less of a threat in warfare than it once was, it still poses a threat to military and civilian personnel because of current concerns for its deployment in a terrorist attack.
  • Sulfur mustards are classic vesicating agents that mainly affect the skin, eyes, and respiratory system. Medical surveillance of individuals exposed to mustard gas in the early 1980's has documented a number of respiratory conditions including bronchiolitis obliterans, asthma, and lung fibrosis that can persist throughout the victims' lifetime.
  • There is currently no known antidote for SM poisoning. Upon exposure, the best recourse is decontamination and supportive treatment. Decontamination of the skin is relatively straight forward and beneficial, whereas internal exposure such a inhalation of sulfur mustards is much more difficult to treat.
  • McClintock et al. report in J. Applied Toxicology, 26(2), 2006, 126-131 that 2-chloroethyl ethyl sulfide (CEES) induced injury of rat lungs can be diminished by the presence of antioxidant enzymes, such as superoxide dismutase (SOD) and/or catalase, delivered via liposomes.
  • WO-A-02078670 discloses the treatment of patients who have suffered damage to the cerebrospinal tissue or who have risk of damage thereto comprising injecting a perfusion fluid in the cerebrospinal pathway containing a neuroprotectant, which may be a free radical inhibitor, including metalloporphyrins, including AEOL10150. The damage can be ischemia, haemorrhage, trauma or exposure to toxic chemicals, such as chemical warfare agents, including the nerve agents VX and Sarin.
  • WO-A-02060383 describes that metalloporphyrins, including 10158, are useful for prevention of toxicity resulting from free radicals produced by chemotherapeutic agents, such as bleomycin, cisplatin, adriamycin (doxorubicin) and camptothicen.
  • It can be seen from the foregoing discussion that there is a need for developing agents that are capable of attenuating, preventing, and/or rescuing organ injury from the deleterious effects resulting from exposure to alkylating agents (e.g., inhalation damage), such as sulfur mustards. The invention addresses these and other needs in the art.
  • BRIEF SUMMARY OF THE INVENTION
  • Disclosed herein are, inter alia, methods for rescuing or preventing organ injury following exposure to alkylating agents by using a substituted porphyrin as the active agent or alkylating agent protectant. The methodology of the invention may implemented as follows.
  • According to one aspect of the invention, there is provided a compound for use in treating an injury associated with exposure to an alkylating-agent, or for use in protecting a subject from the toxic effects associated with exposure to said alkylating agent, the compound having the formula
    Figure imgb0001
    The above compound is also known as "AEOL 10150".
  • The injury may be associated with skin or lungs. The alkylating agent may be a sulfur mustard or 2- chloroethyl ethyl sulfide. Specifically, the alkylating agent is a sulfur mustard. Exposure to the alkylating agent may produce mitochondrial dysfunction, which in turn may result in an increase in reactive oxygen species production or oxidative stress. In particular, exposure to the alkylating agent, relative to non-exposure to the alkylating agent causes an increase in lactate dehydrogenase (LDH) levels, an increase in IgM levels, a decrease of glutathione levels, and an increase in myleperoxidase levels.
  • The compound may be administered by inhalation administration or topical administration. The compound may be administered to the subject within about 0.5 hours to about 48 hours after exposure to the alkylating agent. More specifically, the compound may be administered to the subject within about 1 hour to about 10 hours after exposure to the alkylating agent.
  • Additional features, advantages, and embodiments of the invention may be set forth or apparent from consideration of the following detailed description, and claims. Moreover, it is to be understood that both the foregoing summary of the invention and the following detailed description are exemplary and intended to provide further explanation.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The accompanying drawings, which are included to provide a further understanding of the invention, are incorporated in and constitute a part of this specification, illustrate embodiments of the invention and together with the detailed description serve to explain the principles of the invention. No attempt is made to show structural details of the invention in more detail than may be necessary for a fundamental understanding of the invention and various ways in which it may be practiced.
    • FIGURE 1 shows the structures of bis(2-chloroethyl sulfide), known as SM, and its analog chloroethyl ethyl sulfide (CEES).
    • FIGURE 2 is a graph showing that CEES exposure caused a concentration-dependent injury of human airway epithelial cells. Human lung 16HBE cells were grown to approximately 90% confluence and treated with concentrations of CEES ranging from 600 to 1000 µM for 24 h. Cell viability decreased in a dose-dependent manner as measured by quantifying calcein AM fluorescence. Data represented as mean ± S.E.M., n = 4 where control group fluorescence was defined as 100% viability.
    • FIGURE 3A-3C are graphs showing that CEES exposure produced increased levels of mitochondrial ROS dysfunction. SAE cells (Panel A) and 16HBE cells (Panel B) were treated with 900 µM CEES for 2, 4, 6, 8, 12, 24, and 48 h, after which cells were incubated with the mitochondrial ROS probe MitoSOX (Panel A and Panel B) for 1 h. (Panel C) 16 HBE cells were incubated with the mitochondrial membrane potential indicator Rhodamine 123 for 30 min. MitoSOX fluorescence correlated with increased ROS, where Rhodamine 123 fluorescence was inversely correlated with mitochondrial membrane potential.
    • FIGURE 4 shows chemical structures the catalytic antioxidant metalloporphyrins tested in specific examples 1-6, below.
    • FIGURE 5 is a graph showing the protective effects of metalloporphyrins on CEES-induced cell injury. 16HBE cells were grown to 90% confluence and exposed to 900 µM CEES for a total of 24 h. Cells were treated 1 h after the initial CEES exposure with AEOL 10150, AEOL 10113, AEOL 10303, or MnTBAP at a final concentration of 50 µM in the presence (black bars) or absence (white bars) of 900 µM CEES. Data represented as mean ± S.E.M., n =4. *** , p<0.001 compared with CEES-only treatment group.
    • FIGURE 6A-D are graphs showing the rescue effect of AEOL 10150 on CEES-induced cell death. SAE cells (Panel A and Panel B) and 16HBE cells (Panel C and Panel D) were exposed to 900 µM CEES with AEOL 10150 at 10, 25, and 50 µM concentrations added 1 h after CEES exposure. Cell viability was measured using both calcein AM (Panel A and Panel C) and MTT (Panel B and Panel D) staining with control values being defined as 100% viability. Data represented as mean ± S.E.M., n = 4. **, p <0.01; *** p < 0.001 compared with CEES-only treated group.
    • FIGURE 7A-C are graphs showing that AEOL 10150 rescues CEES-induced increases in mitochondrial ROS and dysfunction. SAE cells (Panel A) and 16HBE cells (Panel B) were exposed to 900 µM CEES for 12 h. AEOL 10150 (50 µM) was added 1 h after CEES exposure. Panel C, 16HBE cells were exposed similar as before except for 4 h. Mitochondrial membrane potential was determined using Rhodamine 123, where fluorescence is inversely correlated with mitochondrial membrane potential. Mean fluorescence was normalized to control levels with controls being 100%. Data represents mean ± S.E.M., n =3 to 6; *, p <0.05; ***, p <0.001 compared with control values. Two-way ANOVA of AEOL 10150, p =0.0563; CEES, p = 0.0033; interaction, p=0.042 (A); AEOL 10150, p =0.1073; CEES, p =0.0004; interaction, p =0.0001 (B); and AEOL 10150, p =0.2876; CEES, p =0.0007; interaction, p = 0.0051 (C).
    • FIGURE 8A-B are graphs showing the effects of CEES on markers of cellular oxidative stress and prevention by AEOL 10150 in 16 HBE cells. Panel A: cells exposed to 900 µM CEES for 12 h had decreased total cellular GSH levels, and AEOL 10150 (50 µM) rescued this decrease when treated 1 h after CEES exposure. Total GSH levels were normalized to the amount of protein and expressed as nanomoles of GSH per milligram of protein. Panel B: CEES also increased the levels of the DNA oxidation marker 80HdG, and AEOL 10150 (50 µM) post-CEES treatment decreased the levels of DNA oxidation. Data expressed as a ratio of 80HdG per 105 2dG. Data presented as mean ± S.E.M., n =4 to 8; *, p <0.05; ***, p < 0.001 compared with control levels. Panel A: two-way ANOVA of AEOL 10150, p =0.1444; CEES, p =0.0001; interaction, p= 0.0481; Panel B: two-way ANOVA of AEOL 10150, p =0.1394; CEES, p =0.0001; interaction, p = 0.0004.
    • FIGURE 9A-D are graphs showing the effects of CEES on markers of injury, edema and inflammation and prevention by AEOL 10150 in rat lung. Panel A: the cytotoxicity marker lactate dehydrogenase (LDH) was measured spectrophotometrically. Panel B: protein levels which are a marker for edema were measured and was measured spectrophotometrically. Panel C: IgM, which is a marker of lung leak was measured by ELISA. Panel D: BAL cells, which are a marker of inflammation and hemorrhage were measure differential cytometry.
    • FIGURE 10 is a graph showing LDH levels in the BAL were increased as a result of CEES inhalation; these levels were decreased to control values when AEOL 10150 was given following CEES. Levels of LDH in the BAL leak were significantly increased as a result of CEES, indicative of epithelial damage and thus leak from those damaged cells. Post exposure treatment with AEOL 10150 significantly decreased LDH leak from cells. Data are shown as mean ± S.E.M., protein n=5 to 9. **, p< 0.01;***, p< 0.001.
    • FIGURE 11A-B are graphs showing the protective effect of AEOL 10150 on CEES-induced increases in BAL protein levels and BAL IgM. At 1 and 9 hours following CEES exposure, rats were treated with AEOL 10150 (5 mg/kg, SC). At 18 hours post exposure, rats were lavaged and levels of BAL protein and IgM were measured. Panel A: CEES exposure resulted in significant increases in BAL protein, while AEOL 10150 treatment with CEES exposure resulted in a significant decrease in protein in the BAL. Panel B: shows a significant increase in BAL IgM as a result of CEES exposure and a subsequent significant decrease in BAL IgM with AEOL 10150 treatment following CEES exposure. Data are shown as mean ± S.E.M., protein n=6 to 16. ***, p< 0.001. IgM n=6. ***, p< 0.001.
    • FIGURE 12A-C are graphs showing that CEES inhalation resulted in increases in BAL RBCs and PMN; treatment with AEOL 10150 reduced BAL RBCs and PMN in BAL. Panel A: In EtOH+PBS or EtOH+AEOL 10150 treated rats, there were very low levels of RBCs. In the CEES+PBS group, rats had significantly increased RBCs in the BAL, indicative of hemorragic injury. Panel B: Neutrophils (polymorphonuclear cells, PMN) were also significantly increased in CEES+PBS treated rats as compared to both EtOH treatment groups. Treatment with AEOL 10150 following CEES resulted in significant decreases in PMN as compared to CEES+PBS. Macrophages were not significantly changed in any of the treatment groups. Data are mean ± S.E.M., n=6 to 13. *,p=0.05; **, p< 0.01;***, p< 0.001.
    • FIGURE 13 is a graph showing that lung tissue myeloperoxidase levels were significantly increased in the CEES+PBS group; treatment with AEOL 10150 significantly decreased lung myeloperoxidase levels as compared to CEES+PBS. Lung tissue was perfused and snap frozen at the time of euthanization. Lung tissue was homogenized in HTAB buffer. Oxidation of tetramethylbenzidine (TMB) was followed for 3 minutes; this data was used to calculate a rate of change. An extinction coefficient for TMB of 3.9 x 104 M-1 cm-1 at 652 nm was used to calculate Units of peroxidase activity and activity was normalized to protein levels using the BCA protein assay. Data are shown as mean ± S.E.M., n=6. *, p=0.05; **, p< 0.01.
    • FIGURE 14 is a graph showing that the DNA oxidation marker 8-hydroxydeoxyguanosine (8-OHdG) was significantly increased as a result of CEES inhalation; treatment with AEOL 10150 significantly decreased CEES-induced DNA oxidation. Data are shown as mean ± S.E.M., n=12. *, p=0.05; **, p< 0.01.
    • FIGURE 15 is a graph showing that levels of the lipid peroxidation marker 4-hydroxynonenal (4-HNE) were elevated as a result of CEES exposure, treatment with AEOL 10150 significantly decreased levels of 4-HNE. Data are shown as mean ± S.E.M., n=11 for EtOH+PBS and CEES+PBS, n= 5 for EtOH+10150 and CEES+10150. *, p=0.05; **.
    DETAILED DESCRIPTION OF THE INVENTION
  • It is be noted that as used herein and in the appended claims, the singular forms "a," "an," and "the" include the plural reference unless the context clearly dictates otherwise. Thus, for example, a reference to "a cell" is a reference to one or more cells.
  • Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art to which the invention pertains. The embodiments of the invention and the various features and advantageous details thereof are explained more fully with reference to the embodiments and examples that are described and/or illustrated in the accompanying drawings and detailed in the following description. It should be noted that the features illustrated in the drawings are not necessarily drawn to scale. Descriptions of well-known components and processing techniques may be omitted so as to not unnecessarily obscure the embodiments of the invention. The examples used herein are intended merely to facilitate an understanding of ways in which the invention may be practiced and to further enable those of skill in the art to practice the embodiments of the invention.
  • Accordingly, provided immediately below is a "Definition" section, where certain terms related to the invention are defined specifically for clarity, but all of the definitions are consistent with how a skilled artisan would understand these terms.
    • SM is sulfur mustard
    • CEES is 2-chloroethyl ethyl sulfide
    • SOD is superoxide dismutase
    • ROS is reactive oxygen species
    • RNS is reactive nitrogen species
    • GSH is glutathione
    • 80HdG is 8-hydroxydeoxyguanosine
    • MTT is 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
    • ANOVA is analysis of variance
    • HBE is human bronchiolar epithelial cells
    • SAEC is human small airway epithelial cells
    • 4-HNE is 4-hydroxynonenal
  • "Alkylating agent," as used herein, generally refers to compounds containing alkyl groups that readily attach to other molecules thereby forming a covalent bond. This process may also be referred to as alkylation. Generally, alkylating agents can disrupt DNA function through different mechanisms, such as: (i) by alkylating DNA bases, thereby preventing DNA synthesis and RNA transcription, (ii) by mediating the formation of cross-bridges, bonds between atoms in the DNA strand, or (iii) by facilitating the mispairing of the nucleotides in the DNA strand thereby leading to mutations. Also, alkylating agents may initiate oxidative stress within the cells of the exposed organ system causing an overall decrease in intracellular glutathione (GSH) and increased DNA oxidation. Exposure to alkylating agents may cause blistering of the skin, damage to the eyes, and damage to the respiratory tract. Exposure to alkylating agents may also cause systemic toxic effects, such as nausea and vomiting, reduction in both leukocytes and erythrocytes, hemorrhagic tendencies, edema, depletion of glutathione, increased myleperoxidase (MPO), increased lactate dehydrogenase (LDH), and increased IgM.
  • "Oxidation," as used herein, is a chemical reaction that transfers electrons from a substance to an oxidizing agent. Oxidation reactions may produce free radicals, which result in oxidative stress and may ultimately result in cell death.
  • "Reactive oxygen species," as used herein, generally refers to free radicals, reactive anions containing oxygen atoms, or molecules containing oxygen atoms that can either produce free radicals or are chemically activated by them. Reactive oxygen species may include, without limitation, superoxide radicals, hydrogen peroxide, peroxynitrite, lipid peroxides, hydroxyl radicals, thiyl radicals, superoxide anion, organic hydroperoxide, RO• alkoxy and ROO• peroxy radicals, and hypochlorous acid. The main source of reactive oxygen species (ROS) in vivo is aerobic respiration, although reactive oxygen species are also produced by peroxisomal b-oxidation of fatty acids, microsomal cytochrome P450 metabolism of xenobiotic compounds, stimulation of phagocytosis by pathogens or lipopolysacchrides, arginine metabolism, tissue specific enzymes. Accumulating oxidative damage may also affect the efficiency of mitochondria and further increase the rate of ROS production.
  • "Reactive nitrogen species," as used herein, generally refers to a family of biomolecules derived from nitric oxide (NO•) and may be produced in animals through the reaction of nitric oxide (NO•) with superoxide (O2 -) to form peroxynitrite (ONOO-). In general, reactive nitrogen species act together with reactive oxygen species to damage cells, resulting in nitrosative stress.
  • "Oxidative stress," as used herein, generally refers to cell damage caused by ROS. The primary damage to cells results from the ROS-induced alteration of macromolecules such as polyunsaturated fatty acids in membrane lipids, essential proteins and DNA. As described in U.S. Patent No. 7,189,707 , oxidative stress and ROS have been implicated in a number of disease states such as Alzheimer's disease, cancer, diabetes mellitus, and aging.
  • "Antioxidant," as used herein, generally refers to molecules or compounds with the capability to attenuate or prevent the oxidation of other molecules. Antioxidants may remove free radicals generated from oxidation reaction and inhibit other oxidation reactions by becoming oxidized themselves. Antioxidants may include reducing agents such as thiols or polyphenols. Additionally, antioxidants may include, without limitation, glutathione, vitamin C, vitamin E, catalase, superoxide dismutase, glutathione peroxidase, various other peroxidases, the substituted porphyrin compounds of the invention and any other molecule or compound that is capable of scavenging reactive oxygen species known in the art.
  • "Rescue," as used herein, is generally defined as counteracting, recovering, or conferring protection from the deleterious effects of reactive oxygen species and other free radicals in a subject, organ, tissue, cell, or biomolecule.
  • "Organ," as used herein, generally refers to a tissue that performs a specific function or group of functions within an organism. An exemplary list of organs includes lungs, heart, blood vessels, blood, salivary glands, esophagus, stomach, liver, gallbladder, pancreas, intestines, rectum, anus, endocrine glands such as hypothalamus, pituitary or pituitary gland, pineal body or pineal gland, thyroid, parathyroids, adrenals, skin, hair, nails, lymph, lymph nodes, tonsils, adenoids, thymus, spleen, muscles, brain, spinal cord, peripheral nerves, nerves, sex organs such as ovaries, fallopian tubes, uterus, vagina, mammary glands, testes, vas deferens, seminal vesicles, prostate, and penis, pharynx, larynx, trachea, bronchi, diaphragm, bones, cartilage, ligaments, tendons, kidneys, ureters, bladder, and urethra.
  • "Organ system," as used herein, generally refers to a group of related organs. Organ systems include, without limitation, circulatory system, digestive system, endocrine system, integumentary system, lymphatic system, muscular system, nervous system, reproductive system, respiratory system, skeletal system, and urinary system.
  • "Biomarker," as used herein, generally refers to an organic biomolecule which is differentially present in a sample taken from a subject of one phenotypic status (e.g., exposure to an alkylating agent) as compared with another phenotypic status (e.g., no exposure to an alkylating agent). A biomarker is differentially present between different phenotypic statuses if the mean or median expression level of the biomarker in the different groups is calculated to be statistically significant. Common tests for statistical significance include, among others, t-test, ANOVA, Kruskal-Wallis, Wilcoxon, Mann-Whitney and odds ratio. Biomarkers, alone or in combination, provide measures of relative risk that a subject belongs to one phenotypic status or another. As such, they are useful as markers for disease (diagnostics), therapeutic effectiveness of a drug (theranostics), and for drug toxicity.
  • "Subject," as used herein, includes individuals who require intervention or manipulation due to a exposure or potential exposure to an alkylating agent that can facilitate organ injury. Furthermore, the term "subject" includes non-human animals and humans.
  • "Active agent," as used herein, generally refers to any compound capable of inducing a change in the phenotype or genotype of a cell, tissue, organ, or organism when contacted with the cell, tissue, organ, or organism. For example, the compound may have the ability to scavenge ROS, prevent or attenuate oxidative stress, and protect organs and organ systems from injury due to exposure to an alkylating agent. The compound may include any substituted porphyrin compounds of the invention, such as a superoxide mimetic, a catalase mimetic or a mimetic having both features.
  • A "pharmaceutically acceptable carrier," as used herein, generally refers to pharmaceutical excipients, for example, pharmaceutically, physiologically, acceptable organic or inorganic carrier substances suitable for enteral or parenteral application that do not deleteriously react with the active agent.
  • Where substituent groups are specified by their conventional chemical formulae, written from left to right, they equally encompass the chemically identical substituents that would result from writing the structure from right to left, e.g., -CH2O- is equivalent to -OCH2-.
  • "Effective dose" or "pharmaceutically effective dose," as used herein, generally refers to the amount of the substituted porphyrin(s) described herein that produces a desired therapeutic effect, such as counteracting the deleterious effects of alkylating agent exposure. The precise amount of the effective dose of a such a compound will yield the most effective results in terms of efficacy of treatment in a given subject will depend upon the activity, pharmacokinetics, pharmacodynamics, and bioavailability of a particular substituted porphyrin of the invention, physiological condition of the subject, the nature of the pharmaceutically acceptable carrier in a formulation, and a route of administration, among other potential factors. Those skilled in the clinical and pharmacological arts will be able to determine these factors through routine experimentation consisting of monitoring the subject and adjusting the dosage. Remington: The Science and Practice of Pharmacy (Gennaro ed. 20.sup.th edition, Williams & Wilkins PA, USA) (2000).
  • Methods
  • In one aspect, methods are disclosed for treating, rescuing and/or protecting organ and organ systems in a subject from the deleterious effects resulting from exposure to alkylating agents using substituted porphyrins. A method for treating an injury associated with exposure to an alkylating agent in a subject may include administering to a subject in need thereof an effective amount of AEOL10150. A method for protecting a subject from the toxic effects associated with exposure to an alkylating agent may include administering prophylactically to a subject in need thereof an effective amount AEOL10150. A method is disclosed for rescuing or protecting organ injury by administering AEOL10150 as the active agent of an alkylating agent protectant.
  • Alkylating Agents
  • Alkylating agents contain alkyl groups that combine readily, typically through covalent bonding, with other molecules. Alkylating agents can disrupt DNA function by three mechanisms: (i) alkylating DNA bases, thereby preventing DNA synthesis and RNA transcription, (ii) mediating the formation of cross-bridges, bonds between atoms in the DNA strand, or (iii) facilitating the mis-pairing of the nucleotides in the DNA strand resulting in mutations in the DNA strand. Also, alkylating agents may initiate oxidative stress within the cells of the exposed organ system causing an overall decrease in intracellular glutathione (GSH) and increased DNA oxidation.
  • Sulfur mustard (2, 2'-dichloro diethyl sulfide) is a known potent vessicating agent and inhalation results in apoptosis and necrosis of the airway epithelium, inflammation, edema, and pseudomembrane formation. 2-chloroethyl ethyl sulfide (CEES, half mustard) is a monofunctional analog of SM that can be utilized to elucidate the mechanisms of injury and as an initial screening of therapeutics. Both SM and CEES ( Figure 1 ) are alkylating agents capable of binding macromolecules including proteins, DNA and lipids.
  • Oxidative stress plays a significant role in SM/CEES mediated damage. For example, exposure to CEES causes an imbalance in production of ROS/RNS and antioxidant defenses in favor of the former. There are many factors that contribute to the increase in ROS following SM/CEES exposure. For example, exposure to SM/CEES facilitates the proliferation of inflammatory cells such as polymorphonuclear leukocytes (PMN), which in turn produces oxidants, including superoxide and hypochlorous acid (HOCl). Furthermore, exposure to CEES also results mitochrondrial dysfunction which further drives increased ROS production, and ultimately, oxidative stress.
  • As discussed above, following exposure to SM/CEES, there is irreparable damage to the respiratory tract such as apoptosis and necrosis of the airway epithelium. However, in certain embodiments of the invention, administration of the substituted porphyrins of the invention subsequent to alkylating agent exposure, have been shown to significantly improve the outcome. For example, administration of AEOL 10150 following CEES exposure have been shown to rescue lung cells and airway cells from alkylating agent-induced toxicity, prevent alkylating agent-mediated ROS and dysfunction, and alkylating agent-induced oxidative stress. AEOL 10150 has been shown to reduce alkylating agent-induced cytotoxicity, reduce alkylating agent-induced increases of protein and IgM in the lung, reduce levels of RBCs and inflammatory cells in the lung, decrease tissue accumulation of PMN, and prevent alkylating agent-induced oxidative stress.
  • The compound may be administered prophylactically to serve as a protectant against exposure to an alkylating agent. The compound may be administered in the dosage amounts specified above about 1 hour to about 48 hours prior to alkylating agent exposure. In specific embodiments, the compound of the invention may be administered about 1 to about 24 hours, more specifically, about 1 to about 12 hours, more specifically about 1 to about 6 hours, and even more specifically, about 1 to about 6 hours prior to alkylating agent exposure.
  • For any compound described herein, the therapeutically effective amount can be initially determined from cell culture assays. Target concentrations will be those concentrations of active compound(s) that are capable of counteracting the effects of the alkylating agent, by monitoring the presence, absence, or alteration in levels of the biomarkers indicative of alkylating agent exposure, such as glutathione, LDH, IgM, and 80-HdG, for example. Methods for measuring the levels of such compounds is known by those of skill in the art and is matter of routine experimentation.
  • Therapeutically effective amounts for use in humans may be determined from animal models. For example, a dose for humans can be formulated to achieve a concentration that has been found to be effective in animals. The dosage in humans can be adjusted by monitoring the levels of the biomarkers indicative of exposure to an alkylating agent and adjusting the dosage upwards or downwards.
  • EXAMPLES
  • For the purpose of the following specific examples, the compounds of Formulas IV-IX, will be designated as indicated in Table 1, below.
    Figure imgb0002
    Figure imgb0003
    Table 1
    Compound of Formula AEOL No. Designation
    IV AEOL 10153
    V AEOL 10158
    VI AEOL 10123
    VII AEOL 10150
    VIII AEOL 10151
    IX AEOL 10303
    X AEOL 10113
    Compounds AEOL 10153, AEOL 10158, AEOL 10123, AEOL 10151, AEOL 10303 and AEOL 10113 are included in the examples for the purpose of reference and fall outside the scope of the claimed invention.
  • Specific Example 1: CEES-Induced Airway Epithelial Cell Injury
  • Human lung 16HBE cells were grown to approximately 90% confluence and treated with increasing concentrations of CEES, ranging from about 600 to about 000 µM. Cell viability was determined by measuring the fluorescence of calcein AM and was found to decrease in a dose-dependent manner from 80% with the 600 µM CEES to below 10% with 1000 µM CEES (Figure 2). 900 µM CEES was used as the optimal dose to carry out the cytoprotection studies because it provided enough cell injury (about 50%) for potential therapeutics to demonstrate efficacy and the most consistent cell injury response in the two cell systems. Because of observed increased resistance of SAE cells to CEES toxicity as seen with 16HBE cells, these exposures were prolonged to 48 h in the SAE cells to provide similar injury responses for comparison of antioxidant protective effects between cell systems.
  • Specific Example 2: Delayed Increase in Mitochondrial ROS and Dysfunction with CEES Exposure
  • As discussed above, mitochondria are a major source of cellular ROS production. Both SAE and 16HBE cells were exposed to 900 µM CEES for 2, 4, 6, 8, 12, 24, and 48 h, after which the cells were incubated with MitoSOX (MitoSOX is a mitochrondrially targeted ROS probe) and fluorescence was measured using flow cytometry. CEES exposure increased ROS levels that peaked at 12 h, and this time-dependent increase was seen in both SAE ( Figure 3A ) and 16HBE ( Figure 3B ) cells. As a consequence, further exposure studies measuring markers of cellular stress were examined after 12 h of exposure.
  • Next CEES was examined to determine whether CEES exposure was associated with any mitochondrial dysfunction. Mitochondria need to maintain a membrane potential to actively make ATP. To examine this, measured Rho 123 fluorescence was measured, which is inversely correlated with mitochondrial membrane potential. Human lung 16HBE cells were exposed to CEES for 2, 4, 6, 8, 12, 24, and 48 h, after which the cells were incubated with Rho 123, and fluorescence was measured using flow cytometry. The results showed that CEES produced a decrease in mitochondrial membrane potential by 4 h, which persisted for 24 h as evidenced by the increase in Rho 123 fluorescence ( Figure 3C ). Notably, there was a significant decrease in Rho 123 fluorescence at 48 h, which can be attributed to the cell death that would be expected to occur based on previous cell viability tests.
  • Specific Example 3: Metalloporphyrins Rescue Human Lung Cells from CEES-Induced Toxicity
  • Several structurally different metalloporphyrins (AEOL 10150, AEOL 10113, AEOL 10303, and MnTBAP) were screened in 16HBE cells for efficacy against CEES toxicity 1 h after the initial exposure ( Figure 4 ). Cells were treated with CEES for 1 h at 37°C, after which the compounds of Formula 10150 (Formula VI, above), 10113 (Formula IX, above), 10103 (Formula VIII, above) and MnTBAP were added at a final concentration of 50 µM. After 24 h, cell viability was measured using calcein AM fluorescence. Three catalytic antioxidant compounds significantly increased cell viability in CEES-exposed cells to 60, 56, and 41% in the 10150, 10113, 10103 groups compared with only 20% in CEES-only exposed cells ( Figure 5 ). Of the four compounds tested, only MnTBAP did not show any protection.
  • Specific Example 4: AEOL 10150 Rescues Human Primary Airway Cells from CEES-Induced Toxicity
  • Primary human lung SAE cells and 16HBE cells were exposed to 900 µM CEES for 48 h. Treatment with AEOL 10150 (10, 25, and 50 µM) occurred 1 h after the initial CEES exposure. AEOL 10150 (50 µM) alone did not change the viability of the cells, as measured by both the calcein AM ( Figure 6 , A and C) and the MTT ( Figure 6 , B and D) assays. CEES alone resulted in a 50% decrease in cell viability, and this was significantly attenuated at the highest concentration of AEOL 10150, to 80% of the control in SAE cells ( Figure 6 , A and B) and nearly 90% in 16HBE cells ( Figure 6 , C and D). Although neither 10 nor 25 µM AEOL 10150 showed a significant increase in viability in the SAE cells, 25 µM AEOL 10150 did show a significant increase in viability in the 16HBE cells. Similar results were obtained in both the calcein AM and the MTT assays used to assess cell viability.
  • Specific Example 5: AEOL 10150 Prevents CEES-Mediated Mitochondrial ROS and Dysfunction
  • AEOL 10150 were assessed to determine whether its cytoprotective effects are associated with CEES-mediated changes in mitochondrial ROS and dysfunction. Cells were grown to approximately 90% confluence and exposed to 900 µM CEES with and without AEOL 10150 (50 µM). Cells were incubated with MitoSOX 12 h after CEES exposure, and fluorescence was measured using flow cytometry. AEOL 10150 added 1 h after CEES treatments significantly decreased mitochondrial ROS compared with CEES exposed cells in both SAE ( Figure 7A ) and 16HBE ( Figure 7B ) cells. AEOL 10150 alone did not cause a change in mitochondrial ROS.
  • Additionally, AEOL 10150 was assessed to determine if it can protect the mitochondria from CEES-induced dysfunction. Lung 16HBE cells were exposed to 900 µM CEES for 4 h with 50µM AEOL 10150 added 1 h after the initial CEES exposure. The CEES-only treated groups showed an increase in Rhodamine 123 fluorescence, indicating a significant loss of mitochondrial membrane potential that was attenuated in the AEOL 10150-treated cells ( Figure 7C ).
  • Specific Example 6: AEOL 10150 Prevents CEES-Induced Oxidative Stress
  • Oxidative stress can result from an imbalance between oxidant production and antioxidant defense. As discussed above, GSH is a major cellular antioxidant. So, the effect of CEES on total cellular GSH levels was determined as well as whether AEOL 10150 altered CEES-mediated changes in GSH levels. Human lung 16HBE cells were exposed for 12 h to CEES, and AEOL 10150 (50 µM) was added 1 h post-CEES treatment. AEOL 10150 alone did not alter intracellular GSH levels, whereas CEES caused a significant decrease in intracellular GSH levels ( Figure 8A AEOL 10150 treatment prevented the CEES-induced decrease in GSH, further implicating an imbalance in redox status of the cells caused by CEES that was reversible by AEOL 10150.
  • One consequence of oxidative stress is an increase in the oxidation of cellular macromolecules. A classic marker for DNA oxidation is the formation of 8-hydroxydeoxyguanosine (8O-HdG), which was determined 12 h after CEES exposure. CEES caused a significant increase in 80HdG levels in lung 16HBE cells as measured by high-performance liquid chromatography ( Figure 8B ). Moreover, AEOL 10150 added 1 h post-CEES exposure decreased CEES-mediated DNA oxidation. These data further support the role of oxidative stress in CEES-mediated injury that is ameliorated by the catalytic antioxidant metalloporphyrin, AEOL 10150.
  • Specific Example 7: AEOL 10150 Protects CEES-Induced Lung Injury in Rat
  • Rats were exposed to 5% CEES for 15 minutes and killed 18 hours later. Groups of rats received AEOL 10150 (5 mg/kg sc, bid) 1 hour after CEES exposure. Rat lungs were lavaged and markers of cytotoxicity, inflammation and edema were measured in bronchoalveolar lavage fluid (BALF). As shown in Figure 9 , CEES caused a significant increase in the ROS. Moreover, AEOL 10150 added 1 h post-CEES exposure decreased CEES-mediated DNA oxidation. These data further support the role of oxidative stress in CEES-mediated injury that is ameliorated by the catalytic antioxidant metalloporphyrin, AEOL 10150.
  • Specific Example 8: AEOL 10150 Reduces CEES-induced Cytotoxicity as Measured by LDH Release
  • CEES-induced cytotoxicity may be assessed by measuring LDH release in the lung. LDH release in the bronchoalveolar lavage fluid (BAL) is a marker of cellular injury in the epithelium. Figure 10 shows levels of LDH release were not different between EtOH + PBS and EtOH + AEOL 10150 treated animals. Following CEES exposure with PBS treatment, LDH release doubled as compared to the control groups (p < 0.01). When rats were administered AEOL 10150 following CEES-exposure, LDH levels were significantly attenuated as compared to the CEES + PBS group (p < 0.001).
  • Specific Example 9: AEOL 10150 Reduces CEES-induced BAL Increases in Protein and IgM
  • Administering AEOL 10150 reduces alkylating agent-induced increases in protein and IgM in the lung. BAL in normal rats consists of macrophages and low levels of large proteins such as albumin. Measuring protein levels in the BAL is one way to measure the accumulation of extravascular protein in the airways. As shown in Figure 11A , compared to EtOH + PBS or EtOH + AEOL 10150, protein levels in BAL were significantly increased as a result of 5% CEES + PBS (p < 0.001). Protein levels in the BAL were significantly decreased from CEES + PBS when animals were administered AEOL 10150 (p < 0.001). Although increased protein levels in BAL may not be a clear indicator of vascular permeability because it may also indicate lysis of damaged epithelium resulting from CEES exposure, the presence of very high molecular weight molecules such as IgM (900 kD) are clearly indicative of increased vascular permeability. Accordingly, Figure 11B demonstrates that IgM levels in the BAL were significantly increased a result in CEES + PBS rats as compared to EtOH + PBS or EtOH + AEOL 10150 (p < 0.001). IgM levels were significantly decreased with CEES + AEOL 10150 treatment as compared to CEES + PBS. Combined, these data demonstrate that administration of the AEOL 10150 following CEES exposure decreased protein levels in BAL as well as IgM levels.
  • Specific Example 10: AEOL 10150 Treatment Reduces Levels of RBCs and Inflammatory Cells in BAL
  • Administering AEOL 10150 following alkylating agent exposure reduces levels of red blood cells (RBCs) and inflammatory cells in the lung. RBCs should not be present in the lung in any considerable levels unless there is hemorrhagic injury. Exposure to 5% CEES + PBS results in significantly increased hemorrhage as shown by increased RBC levels in the BAL (p < 0.001). This CEES-induced damage is ameliorated with AEOL 10150 treatment 18 hours after CEES exposure (p < 0.05). Levels of PMN or neutrophils in the BAL were significantly increased in the CEES + PBS rats as compared to EtOH + PBS or EtOH+10150 (p < 0.001). CEES-induced neutrophil increases were significantly decreased with AEOL 10150 treatment (p < 0.05). While there was a decrease in macrophage levels with CEES exposure, this change did not reach significance as compared to the EtOH exposed animals.
  • Specific Example 11: Myeloperoxidase (MPO) in Lung Homogenate
  • MPO is a glycoprotein expressed in all cells of the myeloid lineage but is most abundant in the azurophilic granules ofPMNs. Released MPO by activated PMNs measured in whole lung homogenate demonstrates tissue accumulation and is a useful complement to measurement of PMN in the BAL. MPO levels were significantly increased as a result of CEES+PBS indicating an increase in PMN tissue accumulation (p < 0.01, Figure 12 ). AEOL 10150 treatment after CEES treatment significantly decreased tissue accumulation of PMN (p < 0.05).
  • Specific Example 12: AEOL 10150 Prevents CEES-induced Oxidative Stress
  • Oxidative stress occurs when oxidant production exceeds antioxidant defense. One marker of oxidative damage is DNA oxidation, which can be measured by the formation of 8-hydroxy-2-deoxyguanosine (8OHdG). 8OHdG significantly increased in CEES+PBS rats as compared to levels in EtOH+PBS (p <0.01) or EtOH+ 10150 (p < 0.05) treatment 18 hours after exposure as measured by HPLC ( Figure 13 ). When rats were exposed to CEES and then received AEOL 10150, 8O-HdG levels were significantly decreased as compared to CEES+PBS (p < 0.05). These data further support the role of oxidative stress in CEES-mediated injury that is ameliorated by the catalytic antioxidant metalloporphyrin, AEOL 10150.
  • Another marker of oxidative damage is the formation of lipid peroxidation products including 4-hydroxynonenal (4-HNE). 4-HNE is a major product of total unsaturated aldehydes formed during lipid peroxidation. Measurement of 4-HNE levels in the lung 18 hours after CEES exposure resulted in a significant increase compared with EtOH+PBS treated rats ( Figure 14 ). AEOL 10150 significantly inhibited CEES- induced lipid peroxidation.

Claims (10)

  1. A compound for use in treating an injury associated with exposure to an alkylating agent, or for use in protecting a subject from the toxic effects associated with exposure to said alkylating agent, the compound having the formula
    Figure imgb0004
  2. The compound for use according to claim 1 wherein the alkylating agent is selected from the group consisting of a sulfur mustard and 2-chloroethyl ethyl sulfide.
  3. The compound for use according to claim 1 or claim 2, wherein the compound is for use in administration by inhalation.
  4. The compound for use according to any of claims 1 to 3 wherein the compound is for use in treating lung injury induced by the alkylating agent.
  5. The compound for use according to claim 1 or claim 2 wherein the compound is for use in topical administration.
  6. The compound of use according to any of claims 1, 2 and 5, wherein the compound is for use in treating skin injury induced by the alkylating agent.
  7. The compound for use according to any preceding claim, wherein the injury is associated with exposure to a sulfur mustard.
  8. The compound for use according to any preceding claim, wherein the compound is for use in administration within about 0.5 hours to about 48 hours after exposure to said alkylating agent.
  9. The compound for use according to any preceding claim, wherein the compound is for use in administration within about 1 hour to about 10 hours after exposure to said alkylating agent.
  10. The compound for use according to any preceding claim, wherein the compound is for use in reducing cytotoxicity induced by the alkylating agent.
EP09805313.5A 2008-05-23 2009-05-26 Compounds for use in treating injury associated with exposure to an alkylating species Active EP2300003B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP14154916.2A EP2732817B1 (en) 2008-05-23 2009-05-26 A compound for use in treating injury associated with exposure to phosgene or chlorine gas

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US5591908P 2008-05-23 2008-05-23
PCT/US2009/045198 WO2010016965A2 (en) 2008-05-23 2009-05-26 Methods for treating injury associated with exposure to an alkylating species

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP14154916.2A Division EP2732817B1 (en) 2008-05-23 2009-05-26 A compound for use in treating injury associated with exposure to phosgene or chlorine gas

Publications (3)

Publication Number Publication Date
EP2300003A2 EP2300003A2 (en) 2011-03-30
EP2300003A4 EP2300003A4 (en) 2012-04-04
EP2300003B1 true EP2300003B1 (en) 2014-03-05

Family

ID=41664134

Family Applications (2)

Application Number Title Priority Date Filing Date
EP14154916.2A Active EP2732817B1 (en) 2008-05-23 2009-05-26 A compound for use in treating injury associated with exposure to phosgene or chlorine gas
EP09805313.5A Active EP2300003B1 (en) 2008-05-23 2009-05-26 Compounds for use in treating injury associated with exposure to an alkylating species

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP14154916.2A Active EP2732817B1 (en) 2008-05-23 2009-05-26 A compound for use in treating injury associated with exposure to phosgene or chlorine gas

Country Status (13)

Country Link
US (2) US11382895B2 (en)
EP (2) EP2732817B1 (en)
JP (1) JP5608644B2 (en)
KR (2) KR101646066B1 (en)
CN (1) CN102105146A (en)
AU (1) AU2009280042B2 (en)
CA (1) CA2725012C (en)
DK (1) DK2300003T3 (en)
ES (2) ES2600469T3 (en)
IL (1) IL209435A (en)
MX (1) MX2010012593A (en)
RU (1) RU2506083C2 (en)
WO (1) WO2010016965A2 (en)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2010289385A1 (en) * 2009-09-02 2012-03-15 The Regents Of The University Of Colorado, A Body Corporate Methods of treating mitochondrial disorders using metalloporhyrins
WO2013130150A2 (en) * 2011-12-02 2013-09-06 The Regents Of The University Of Colorado, A Body Corporate Metalloporphyrin neurological treatments
WO2015077627A1 (en) * 2013-11-22 2015-05-28 Aeolus Sciences, Inc. Synthesis and formulations of porphyrin compounds
RU2635504C1 (en) * 2016-12-01 2017-11-13 Войсковая Часть 41598 Benzylpenicillin sodium salt as means of preventive measures and early medical maintenance of acute sulphur mustard damage
RU2629602C1 (en) * 2016-12-01 2017-08-30 Войсковая Часть 41598 Bicillin-3 as means for prevention and early therapy of acute sulphur mustard injuries
RU2635476C1 (en) * 2016-12-02 2017-11-13 Войсковая Часть 41598 Bicillin-1 as means for prevention and early therapy of acute injuries caused by sulphur mustard
WO2020123057A2 (en) * 2018-10-31 2020-06-18 Repurposed Therapeutics, Inc. Dba Defender Treatment of exposure to chlorine gas with scopolamine

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7189707B2 (en) * 1999-01-25 2007-03-13 National Jewish Medical Research Center Substituted porphyrins
US20070149498A1 (en) * 2001-01-19 2007-06-28 Aeolus Sciences, Inc. Cancer therapy

Family Cites Families (119)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2951799A (en) 1957-11-13 1960-09-06 Monsanto Chemicals Photoxidation processes using heterocyclic photosensitizers
DE3464252D1 (en) 1983-06-03 1987-07-23 Hoffmann La Roche Labelled molecules for fluorescence immunoassays and processes and intermediates for their preparation
US4829984A (en) 1983-12-15 1989-05-16 Gordon Robert T Method for the improvement of transplantation techniques and for the preservation of tissue
US4963367A (en) 1984-04-27 1990-10-16 Medaphore, Inc. Drug delivery compositions and methods
US5087438A (en) 1984-07-03 1992-02-11 Gordon Robert T Method for affecting intracellular and extracellular electric and magnetic dipoles
US4911920A (en) 1986-07-30 1990-03-27 Alcon Laboratories, Inc. Sustained release, comfort formulation for glaucoma therapy
GB8429845D0 (en) 1984-11-26 1985-01-03 Efamol Ltd Porphyrins & cancer treatment
US4758422A (en) 1985-01-04 1988-07-19 Salutar Inc. Ferrioxamine paramagnetic contrast agents for MR imaging
SU1287521A1 (en) * 1985-01-10 1996-10-27 Г.Б. Маравин 2,7,12,18-tetramethyl-3,8-diethyl-13,17-di-(3-dimethylaminopropyl)-porphyrin salts showing antiiradiation activity and a method of their synthesis
US4657902A (en) 1985-03-25 1987-04-14 The Rockefeller University Therapeutic use of tin mesoporphyrin
ATE78158T1 (en) 1985-05-22 1992-08-15 Liposome Technology Inc METHOD AND SYSTEM FOR INHALATION OF LIPOSOMES.
US5248603A (en) 1985-09-03 1993-09-28 Symbicom Aktiebolag Superoxide dismutase
DK402785D0 (en) 1985-09-03 1985-09-03 Syn Tek Ab PROCEDURE FOR THE PREPARATION OF AN ENZYM
FR2588189B1 (en) 1985-10-03 1988-12-02 Merck Sharp & Dohme LIQUID-GEL PHASE TRANSITION PHARMACEUTICAL COMPOSITION
US4866054A (en) 1986-05-13 1989-09-12 Chai-Tech Corporation Antioxidant metallo-organic treatment of inflammation
US4746735A (en) 1986-11-21 1988-05-24 The Dow Chemical Company Regiospecific aryl nitration of meso-substituted tetraarylporphyrins
US5162231A (en) 1989-10-25 1992-11-10 Cole Dean A Method of using 5,10,15,20-tetrakis(carboxyphenyl)porphine for detecting cancers of the lung
DE3854872D1 (en) 1987-03-14 1996-02-22 Boehringer Ingelheim Int Human manganese superoxide dismutase (hMn-SOD)
US5223538A (en) 1987-03-31 1993-06-29 Duke University Superoxide dismutase mimic
US5227405A (en) 1987-03-31 1993-07-13 Duke University Superoxide dismutase mimic
US4892941A (en) 1987-04-17 1990-01-09 Dolphin David H Porphyrins
US4851403A (en) 1987-04-21 1989-07-25 Johnson Matthey, Inc. Radiation sensitizers
US4885114A (en) 1987-04-22 1989-12-05 Barnes Engineering Co. Metallized tetra((meso)-5-methyl-2-thiophene)porphines, platinum (5-bromo octaethylporphine) and optical filters containing same
US5051337A (en) 1987-06-10 1991-09-24 Director-General Of Agency Of Industrial Science And Technology Optical recording material
GB8805849D0 (en) 1988-03-11 1988-04-13 Efamol Holdings Porphyrins & cancer treatment
US5162519A (en) 1988-03-11 1992-11-10 Efamol Holdings Plc Porphyrins and cancer treatment
US5284647A (en) 1988-03-18 1994-02-08 Schering Aktiengesellschaft Mesotetraphenylporphyrin complex compounds, process for their production and pharmaceutical agents containing them
DE3809671A1 (en) 1988-03-18 1989-09-28 Schering Ag PORPHYRINE COMPLEX COMPOUNDS, METHOD FOR THE PRODUCTION THEREOF AND PHARMACEUTICAL AGENTS CONTAINING THEM
US5192788A (en) 1988-05-23 1993-03-09 Georgia State University Foundation, Inc. Porphyrin antiviral compositions
US5109016A (en) 1988-05-23 1992-04-28 Georgia State University Foundation, Inc. Method for inhibiting infection or replication of human immunodeficiency virus with porphyrin and phthalocyanine antiviral compositions
FR2632187B1 (en) 1988-06-02 1990-09-14 Centre Nat Rech Scient METALLOPORPHYRIN DERIVATIVES, THEIR PREPARATION, THEIR THERAPEUTIC APPLICATION AND THEIR USE FOR THE PREPARATION OF HYBRID MOLECULES
US5171680A (en) 1988-06-14 1992-12-15 Chiron Corporation Superoxide dismutase analogs having novel binding properties
JPH0746212B2 (en) 1988-12-03 1995-05-17 工業技術院長 Method for producing anisotropic LB film
WO1990010694A1 (en) 1989-03-06 1990-09-20 Suntory Limited New superoxide dismutase
DK455789D0 (en) 1989-09-15 1989-09-15 Symbicom Ab POLYPEPTIDE
US5010073A (en) 1989-10-05 1991-04-23 The Rockefeller University Use of liposomes as carriers for metalloporphyrins
EP0424033A3 (en) 1989-10-19 1991-07-31 Pola Chemical Industries Inc External skin preparation
JPH03273082A (en) 1990-03-20 1991-12-04 Dai Ichi Seiyaku Co Ltd Peroxide decomposer
US5236915A (en) 1990-05-31 1993-08-17 Health Research, Inc. Meso poly(4-sulfonatophenyl) porphines as MRI image enhancing agents
EP0462836A3 (en) 1990-06-20 1992-08-26 Mitsui Toatsu Chemicals, Inc. Recombinant vector plasmid capable of expressing human manganese superoxide dismutase, and process of producing this enzyme
US5202317A (en) 1990-09-13 1993-04-13 The Regents Of The University Of California Synthetic drug molecules that mimic metalloenzymes
US5217966A (en) 1990-09-13 1993-06-08 The Regents Of The University Of California Synthetic drug molecules that mimic metalloenzymes
AU8947791A (en) 1990-11-01 1992-05-26 Scripps Research Institute, The Glycosaminoglycan-targeted fusion proteins, their design, construction and compositions
EP0557417A4 (en) 1990-11-14 1994-06-08 Uab Research Foundation Compositions for reducing oxidative injury
US5192757A (en) 1990-12-20 1993-03-09 Glaxo Inc. Cobalt porphyrins
DK0495421T3 (en) 1991-01-15 1996-12-09 Alcon Lab Inc Use of carrageenans in topical ophthalmic compositions
GB9103991D0 (en) 1991-02-26 1991-04-10 Nat Res Dev Molecular electronic devices
US5212162A (en) 1991-03-27 1993-05-18 Alcon Laboratories, Inc. Use of combinations gelling polysaccharides and finely divided drug carrier substrates in topical ophthalmic compositions
FR2676738B1 (en) 1991-05-22 1995-05-05 Ir2M NOVEL ORGANIC TRANSITION METAL WITH PORPHYRINIC STRUCTURE, THERAPEUTIC COMPOSITION CONTAINING SAME, IN PARTICULAR WITH HYPOGLYCEMIC ACTIVITY.
AU661023B2 (en) 1991-07-19 1995-07-13 Monsanto Company Manganese complexes of nitrogen-containing macrocyclic ligands effective as catalysts for dismutating superoxide
CA2072934C (en) 1991-07-19 2007-08-28 Karl William Aston Manganese complexes of nitrogen-containing macrocyclic ligands effective as catalysts for dismutating superoxide
US5262532A (en) 1991-07-22 1993-11-16 E.R. Squibb & Sons, Inc. Paramagnetic metalloporphyrins as contrast agents for magnetic resonance imaging
US5212300A (en) 1991-09-12 1993-05-18 Sun Company, Inc. (R&M) Cyano- and polycyanometallo-porphyrins as catalysts for alkane oxidation
US5976551A (en) 1991-11-15 1999-11-02 Institut Pasteur And Institut Nationale De La Sante Et De La Recherche Medicale Altered major histocompatibility complex (MHC) determinant and method of using the determinant
US5371199B1 (en) 1992-08-14 1995-12-26 Univ Pennsylvania Substituted porphyrins porphyrin-containing polymers and synthetic methods therefor
US5599924A (en) 1992-08-14 1997-02-04 Trustees Of The University Of Pennsylvania Electron-deficient porphyrins and processes and intermediates for preparing same
US5493017A (en) 1992-08-14 1996-02-20 The Trustees Of The University Of Pennsylvania Ring-metalated porphyrins
ATE201595T1 (en) 1992-09-03 2001-06-15 Univ California METALPORPHYRIN COMPOSITIONS
US5403834A (en) 1992-12-07 1995-04-04 Eukarion, Inc. Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease
US5696109A (en) 1992-12-07 1997-12-09 Eukarion, Inc. Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease
US5834509A (en) 1992-12-07 1998-11-10 Eukarion, Inc. Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease
US6204259B1 (en) 1993-01-14 2001-03-20 Monsanto Company Manganese complexes of nitrogen-containing macrocyclic ligands effective as catalysts for dismutating superoxide
DE4305523A1 (en) 1993-02-17 1994-08-18 Diagnostikforschung Inst Meso-tetraphenylporphyrin complexes, processes for their preparation and pharmaceutical compositions containing them
US5428037A (en) 1993-04-09 1995-06-27 Syntex Pharmaceuticals, Ltd. Heterocyclic derivatives in the treatment of Ischaemia and related diseases
JP3273082B2 (en) 1993-06-18 2002-04-08 三和電気工業株式会社 Sleeve holder for optical connector
US6417182B1 (en) 1993-08-25 2002-07-09 Anormed Inc. Pharmaceutical compositions comprising metal complexes
US6127356A (en) 1993-10-15 2000-10-03 Duke University Oxidant scavengers
US5747026A (en) 1993-10-15 1998-05-05 University Of Alabama At Birmingham Research Foundation Antioxidants
US5994339A (en) 1993-10-15 1999-11-30 University Of Alabama At Birmingham Research Foundation Oxidant scavengers
US5488145A (en) 1993-12-23 1996-01-30 Oklahoma Medical Research Foundation 2,4-disulfonyl phenyl butyl nitrone, its salts, and their use as pharmaceutical free radical traps
US5608054A (en) 1993-12-29 1997-03-04 Sun Company, Inc. (R&M) Porphyrins and metal complexes thereof having haloalkyl side chains
US5405369A (en) 1994-01-25 1995-04-11 Medical College Of Ohio Photochemical ablation of gastro-intestinal tissue for augmentation of an organ
US5604199A (en) 1994-03-29 1997-02-18 The Nemours Foundation Method of treating fibrosis in skeletal muscle tissue
WO1995031197A1 (en) 1994-05-13 1995-11-23 Monsanto Company Methods of use for peroxynitrite decomposition catalysts, pharmaceutical compositions therefor
US6245758B1 (en) 1994-05-13 2001-06-12 Michael K. Stern Methods of use for peroxynitrite decomposition catalysts, pharmaceutical compositions therefor
US5563132A (en) 1994-06-21 1996-10-08 Bodaness; Richard S. Two-step cancer treatment method
US6309853B1 (en) 1994-08-17 2001-10-30 The Rockfeller University Modulators of body weight, corresponding nucleic acids and proteins, and diagnostic and therapeutic uses thereof
WO1996009053A1 (en) 1994-09-20 1996-03-28 Duke University Oxidoreductase activity of manganic porphyrins
CA2132690A1 (en) 1994-09-22 1996-03-23 Dean Willis Control and modulation of inflammatory response in humans in need of such control and modulation
US6013241A (en) 1995-01-23 2000-01-11 Schering Aktiengesellschaft Use of porphyrin-complex or expanded porphyrin-complex compounds as an infarction localization diagnosticum
CA2223407C (en) 1995-06-07 2011-02-08 Duke University Oxidant scavengers
WO1997006831A1 (en) 1995-08-17 1997-02-27 Monsanto Company Methods of diagnostic image analysis using metal complexes of nitrogen-containing macrocyclic ligands
AU6896796A (en) 1995-08-17 1997-03-12 Monsanto Company Methods of diagnostic image analysis using bioconjugates of metal complexes of nitrogen-containing macrocyclic ligands
AU700958B2 (en) 1995-08-17 1999-01-14 Monsanto Company Bioconjugates of manganese complexes and their application as catalysts
US5648523A (en) 1995-10-26 1997-07-15 Chiang Long Y Fullerene derivatives as free-radical scavengers
US5948771A (en) 1996-01-31 1999-09-07 The Trustees Of Columbia University In The City Of New York Method for treating heart failure using tetrapyrroles and metallotetrapyrroles
WO1997033588A1 (en) 1996-03-13 1997-09-18 Monsanto Company Iron complexes of nitrogen-containing macrocyclic ligands effective as catalysts for dismutating superoxide
CZ271198A3 (en) 1996-03-13 1999-01-13 Monsanto Company Manganese bioconjugates of iron complexes of macrocyclic ligands containing nitrogen, acting as catalysts for superoxide dismutation
US5756492A (en) 1996-09-09 1998-05-26 Sangstat Medical Corporation Graft survival prolongation with porphyrins
AU6650198A (en) 1997-02-05 1998-08-25 Board Of Regents, The University Of Texas System Porphyrin compounds as telomerase inhibitors
US6180620B1 (en) 1997-06-20 2001-01-30 G.D. Searle & Co. Analgesic methods using synthetic catalysts for the dismutation of superoxide radicals
US6214817B1 (en) 1997-06-20 2001-04-10 Monsanto Company Substituted pyridino pentaazamacrocyle complexes having superoxide dismutase activity
GB9716446D0 (en) 1997-08-05 1997-10-08 Agrevo Uk Ltd Fungicides
AU8749598A (en) 1997-08-26 1999-03-16 Ihara Chemical Industry Co. Ltd. Biarylalkylenecarbamic acid derivatives and bacteriocides for agricultural an d horticultural use
ES2198767T3 (en) 1997-11-03 2004-02-01 Duke University REPLACED PORPHIRINS.
CA2329751C (en) 1998-04-24 2010-10-12 Duke University Substituted porphyrins
AU5229599A (en) 1998-07-23 2000-02-14 Metaphore, Inc. Libraries of polyhydroxamates and their analogs
WO2000023568A2 (en) 1998-10-06 2000-04-27 Albert Einstein College Of Medicine Of Yeshiva University Methods and compositions for decreasing mitochondrial overproduction of reactive oxygen species in cells
ES2240101T3 (en) 1999-05-27 2005-10-16 Monsanto Company MODIFIED BIOMATERIALS WITH DISMUTE SUPEROXIDE MIMETICS.
US6448239B1 (en) 1999-06-03 2002-09-10 Trustees Of Princeton University Peroxynitrite decomposition catalysts and methods of use thereof
US6608050B2 (en) 1999-07-20 2003-08-19 Pharmacia & Upjohn Company Lyophilizate of lipid complex of water insoluble porphyrins
US6372727B1 (en) 1999-10-13 2002-04-16 Uab Research Foundation Metalloporphyrin treatment of neurologic disease
EP1148057A4 (en) 1999-11-30 2002-05-29 Photochemical Co Ltd Nitroimidazole-supporting porphyrin complex
JP3383839B2 (en) 2000-03-13 2003-03-10 奈良先端科学技術大学院大学長 Novel mercapto-substituted imidazolyl porphyrin metal complex monomer, polymer having the same as a repeating unit, and methods for producing them
US6624187B1 (en) 2000-06-12 2003-09-23 Health Research, Inc. Long wave length absorbing bacteriochlorin alkyl ether analogs
EP1320532A1 (en) 2000-06-14 2003-06-25 Duke University Tetrapyrroles
US6403788B1 (en) 2000-07-11 2002-06-11 Eukarion, Inc. Non-genotoxic metalloporphyrins as synthetic catalytic scavengers of reactive oxygen species
US6573258B2 (en) 2000-09-27 2003-06-03 Frontier Scientific, Inc. Photodynamic porphyrin antimicrobial agents
EP1370240A4 (en) * 2001-03-02 2004-09-22 Neuron Therapeutics Inc Neuroprotectants formulations and methods
AU2002344234B2 (en) 2001-05-31 2007-11-08 Miravant Pharmaceuticals, Inc. Metallotetrapyrrolic photosensitizing agents for use in photodynamic therapy
US6566517B2 (en) 2001-06-06 2003-05-20 Brookhaven Science Associates, Llc Metalloporphyrins and their uses as imageable tumor-targeting agents for radiation therapy
DE10240343A1 (en) * 2002-08-27 2004-03-11 Schering Ag Peroxynitrite rearrangement catalysts
AU2005210668A1 (en) * 2004-01-30 2005-08-18 Angiotech International Ag Compositions and methods for treating contracture
US7807825B2 (en) * 2004-02-09 2010-10-05 Duke University Substituted porphyrins
WO2006083381A2 (en) * 2004-12-03 2006-08-10 University Of Maryland, Baltimore Composition for treating sulfur mustard toxicity and methods of using same
US20090234011A1 (en) 2005-04-21 2009-09-17 Goldstein Glenn A N-acetylcysteine amide (nac amide) for the treatment of diseases and conditions associated with oxidative stress
JP5122460B2 (en) * 2005-09-16 2013-01-16 アラーガン、インコーポレイテッド Compositions and methods for intraocular delivery of therapeutic agents
US7947827B2 (en) * 2006-06-30 2011-05-24 State Of Oregon Acting By And Through The State Board Of Higher Education On Behalf Of Oregon State University Pharmaceutical formulation comprising a metaloporphyrin and method for its purification and use
AU2010289385A1 (en) * 2009-09-02 2012-03-15 The Regents Of The University Of Colorado, A Body Corporate Methods of treating mitochondrial disorders using metalloporhyrins

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7189707B2 (en) * 1999-01-25 2007-03-13 National Jewish Medical Research Center Substituted porphyrins
US20070149498A1 (en) * 2001-01-19 2007-06-28 Aeolus Sciences, Inc. Cancer therapy

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MALASSAGNE B ET AL: "The superoxide dismutase mimetic MnTBAP prevents Fas-induced acute liver failure in the mouse", GASTROENTEROLOGY 2001 US, vol. 121, no. 6, 2001, pages 1451 - 1459, ISSN: 0016-5085 *

Also Published As

Publication number Publication date
KR20110017398A (en) 2011-02-21
US20230000830A1 (en) 2023-01-05
CA2725012A1 (en) 2010-02-11
MX2010012593A (en) 2011-03-04
AU2009280042B2 (en) 2015-06-04
KR20160036108A (en) 2016-04-01
CA2725012C (en) 2019-05-07
RU2010152638A (en) 2012-06-27
DK2300003T3 (en) 2014-05-26
EP2300003A2 (en) 2011-03-30
WO2010016965A9 (en) 2010-04-01
EP2732817B1 (en) 2016-08-24
EP2300003A4 (en) 2012-04-04
IL209435A0 (en) 2011-01-31
IL209435A (en) 2015-04-30
KR101646066B1 (en) 2016-08-05
RU2506083C2 (en) 2014-02-10
EP2732817A1 (en) 2014-05-21
ES2600469T3 (en) 2017-02-09
JP2012507471A (en) 2012-03-29
US20110136775A1 (en) 2011-06-09
CN102105146A (en) 2011-06-22
WO2010016965A2 (en) 2010-02-11
JP5608644B2 (en) 2014-10-15
US11382895B2 (en) 2022-07-12
ES2464728T3 (en) 2014-06-03
AU2009280042A1 (en) 2010-02-11

Similar Documents

Publication Publication Date Title
EP2300003B1 (en) Compounds for use in treating injury associated with exposure to an alkylating species
Yu et al. Diallyl trisulfide ameliorates myocardial ischemia–reperfusion injury by reducing oxidative stress and endoplasmic reticulum stress-mediated apoptosis in type 1 diabetic rats: role of SIRT1 activation
Ryter et al. Autophagy in the lung
JP4588325B2 (en) How to treat ileus
Goodman et al. Heme oxygenase-1 protects against radiocontrast-induced acute kidney injury by regulating anti-apoptotic proteins
Millar et al. Effects of iron and iron chelation in vitro on mucosal oxidant activity in ulcerative colitis
EP0327612B1 (en) Pharmaceutical therapeutic use of glutathione derivatives
JP2001513760A (en) Mitochondrial protection method and composition therefor
Pi et al. Melatonin alleviates cardiac dysfunction via increasing Sirt1-mediated beclin-1 deacetylation and autophagy during sepsis
Faller et al. Kinetic effects of carbon monoxide inhalation on tissue protection in ventilator-induced lung injury
Parlar et al. Resveratrol normalizes the deterioration of smooth muscle contractility after intestinal ischemia and reperfusion in rats associated with an antioxidative effect and modulating tumor necrosis factor alpha activity
Zhuang et al. MCTR3 reduces LPS-induced acute lung injury in mice via the ALX/PINK1 signaling pathway
Ahmad et al. Enema based therapy using liposomal formulation of low molecular weight heparin for treatment of active ulcerative colitis: New adjunct therapeutic opportunity
Deniz et al. Radiation-induced oxidative injury of the ileum and colon is alleviated by glucagon-like peptide-1 and-2
Cinci et al. Suppression of allergen-induced respiratory dysfunction and airway inflammation in sensitized guinea pigs by MnII (Me2DO2A), a novel superoxide scavenger compound
Wang et al. Dexmedetomidine alleviates lung oxidative stress injury induced by ischemia-reperfusion in diabetic rats via the Nrf2-Sulfiredoxin1 pathway
Dizaji et al. The effects of Hemiscorpius lepturus induced-acute kidney injury on PGC-1α gene expression: From induction to suppression in mice
Wang et al. Protection of zero-valent iron nanoparticles against sepsis and septic heart failure
Mehany et al. In-vivo study on the rat heart for studying the impact of the antioxidant activity of nanohybrid composite of fluoride-based on chitosan nanoparticles
Choi Cytotoxicity and biokinetic evaluation of clay minerals
JPH08508504A (en) Antioxidant Alkyl Aryl, Polyether Alcohol, Polymers
Wong et al. Case series of paraquat poisoning in Tuen Mun Hospital
Borrelli The use of ozone as redox modulator in the treatment of the chronic obstructive pulmonary disease (COPD)
Norman et al. Intravascular hemolysis associated with severe cutaneous burn injuries in five horses
Chen et al. Exosome-coated polydatin nanoparticles in the treatment of radiation-induced intestinal damage

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20101222

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA RS

DAX Request for extension of the european patent (deleted)
RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 45/06 20060101ALI20120222BHEP

Ipc: A61K 31/409 20060101AFI20120222BHEP

Ipc: A61P 11/00 20060101ALI20120222BHEP

Ipc: A61P 17/00 20060101ALI20120222BHEP

Ipc: A61K 31/4178 20060101ALI20120222BHEP

Ipc: A61K 31/427 20060101ALI20120222BHEP

Ipc: A61K 31/555 20060101ALI20120222BHEP

A4 Supplementary search report drawn up and despatched

Effective date: 20120301

17Q First examination report despatched

Effective date: 20121205

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20130912

RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 45/06 20060101ALI20130830BHEP

Ipc: A61P 17/00 20060101ALI20130830BHEP

Ipc: A61P 39/00 20060101ALI20130830BHEP

Ipc: A61K 31/555 20060101ALI20130830BHEP

Ipc: A61P 11/00 20060101ALI20130830BHEP

Ipc: A61K 31/427 20060101ALI20130830BHEP

Ipc: A61K 31/4178 20060101ALI20130830BHEP

Ipc: A61K 31/409 20060101AFI20130830BHEP

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 654374

Country of ref document: AT

Kind code of ref document: T

Effective date: 20140315

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602009022288

Country of ref document: DE

Effective date: 20140417

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20140523

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: MEYER AND KOLLEGEN, CH

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2464728

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20140603

REG Reference to a national code

Ref country code: NL

Ref legal event code: T3

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

RIN2 Information on inventor provided after grant (corrected)

Inventor name: WHITE, CARL, W.

Inventor name: DAY, BRIAN, J.

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140605

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140305

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140305

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140305

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140305

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140305

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140305

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140605

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140305

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140705

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140305

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140305

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602009022288

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140707

Ref country code: LU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140526

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140305

26N No opposition filed

Effective date: 20141208

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602009022288

Country of ref document: DE

Effective date: 20141208

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140305

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140305

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 8

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140606

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140305

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20090526

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20170526

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20170530

Year of fee payment: 9

Ref country code: DK

Payment date: 20170530

Year of fee payment: 9

Ref country code: CH

Payment date: 20170527

Year of fee payment: 9

Ref country code: IE

Payment date: 20170530

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20170601

Year of fee payment: 9

Ref country code: IT

Payment date: 20170524

Year of fee payment: 9

Ref country code: FI

Payment date: 20170530

Year of fee payment: 9

Ref country code: BE

Payment date: 20170529

Year of fee payment: 9

Ref country code: AT

Payment date: 20170504

Year of fee payment: 9

Ref country code: SE

Payment date: 20170530

Year of fee payment: 9

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140305

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: DK

Ref legal event code: EBP

Effective date: 20180531

Ref country code: SE

Ref legal event code: EUG

Ref country code: NL

Ref legal event code: MM

Effective date: 20180601

REG Reference to a national code

Ref country code: AT

Ref legal event code: MM01

Ref document number: 654374

Country of ref document: AT

Kind code of ref document: T

Effective date: 20180526

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20180526

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20180531

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180526

Ref country code: SE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180527

Ref country code: FI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180526

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180531

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180531

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180601

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180526

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180526

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180526

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180531

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180531

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20190913

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180527

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230424

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20230420

Year of fee payment: 15

Ref country code: DE

Payment date: 20230419

Year of fee payment: 15